51
|
Tanboon J, Nishino I. Autosomal Recessive Limb-Girdle Muscular Dystrophies. CURRENT CLINICAL NEUROLOGY 2023:93-121. [DOI: 10.1007/978-3-031-44009-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
52
|
Savarese M, Jokela M, Udd B. Distal myopathy. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:497-519. [PMID: 37562883 DOI: 10.1016/b978-0-323-98818-6.00002-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Distal myopathies are a group of genetic, primary muscle diseases. Patients develop progressive weakness and atrophy of the muscles of forearm, hands, lower leg, or feet. Currently, over 20 different forms, presenting a variable age of onset, clinical presentation, disease progression, muscle involvement, and histological findings, are known. Some of them are dominant and some recessive. Different variants in the same gene are often associated with either dominant or recessive forms, although there is a lack of a comprehensive understanding of the genotype-phenotype correlations. This chapter provides a description of the clinicopathologic and genetic aspects of distal myopathies emphasizing known etiologic and pathophysiologic mechanisms.
Collapse
Affiliation(s)
- Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland; Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Manu Jokela
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland; Division of Clinical Neurosciences, Department of Neurology, Turku University Hospital, Turku, Finland
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland; Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland; Department of Neurology, Vaasa Central Hospital, Vaasa, Finland.
| |
Collapse
|
53
|
Nashi S, Polavarapu K, Bardhan M, Anjanappa RM, Preethish-Kumar V, Vengalil S, Padmanabha H, Geetha TS, Prathyusha PV, Ramprasad V, Joshi A, Chawla T, Unnikrishnan G, Sharma P, Huddar A, Uppilli B, Thomas A, Baskar D, Mathew S, Menon D, Arunachal G, Faruq M, Thangaraj K, Nalini A. Genotype-phenotype correlation and natural history study of dysferlinopathy: a single-centre experience from India. Neurogenetics 2023; 24:43-53. [PMID: 36580222 DOI: 10.1007/s10048-022-00707-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
Dysferlinopathies are a group of limb-girdle muscular dystrophies causing significant disability in the young population. There is a need for studies on large cohorts to describe the clinical, genotypic and natural history in our subcontinent. To describe and correlate the clinical, genetic profile and natural history of genetically confirmed dysferlinopathies. We analysed a retrospective cohort of patients with dysferlinopathy from a single quaternary care centre in India. A total of 124 patients with dysferlinopathy were included (40 females). Median age at onset and duration of illness were 21 years (range, 13-50) and 48 months (range, 8-288), respectively. The average follow-up period was 60 months (range, 12-288). Fifty-one percent had LGMD pattern of weakness at onset; 23.4% each had Miyoshi and proximo-distal type while isolated hyperCKemia was noted in 1.6%. About 60% were born to consanguineous parents and 26.6% had family history of similar illness. Twenty-three patients (18.6%) lost ambulation at follow-up; the median time to loss of independent ambulation was 120 months (range, 72-264). Single-nucleotide variants (SNVs) constituted 78.2% of patients; INDELs 14.5% and 7.3% had both SNVs and INDELs. Earlier age at onset was noted with SNVs. There was no correlation between the other clinical parameters and ambulatory status with the genotype. Thirty-seven (45.7%) novel pathogenic/likely pathogenic (P/LP) variants were identified out of a total of 81 variations. The c.3191G > A variant was the most recurrent mutation. Our cohort constitutes a clinically and genetically heterogeneous group of dysferlinopathies. There is no significant correlation between the clinico-genetic profile and the ambulatory status.
Collapse
Affiliation(s)
- Saraswati Nashi
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
| | - Mainak Bardhan
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Ram Murthy Anjanappa
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Veeramani Preethish-Kumar
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Seena Vengalil
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Hansashree Padmanabha
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | | | - P V Prathyusha
- Department of Biostatistics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | | | - Aditi Joshi
- CSIR-Institute of Genomics and Integrative Biology (IGIB), Delhi, India
| | - Tanushree Chawla
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Gopikirshnan Unnikrishnan
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Pooja Sharma
- CSIR-Institute of Genomics and Integrative Biology (IGIB), Delhi, India
| | - Akshata Huddar
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | | | - Abel Thomas
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Dipti Baskar
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Susi Mathew
- CSIR-Institute of Genomics and Integrative Biology (IGIB), Delhi, India
| | - Deepak Menon
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Mohammed Faruq
- CSIR-Institute of Genomics and Integrative Biology (IGIB), Delhi, India
| | | | - Atchayaram Nalini
- Department of Neurology, Neuroscience Faculty Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India.
| |
Collapse
|
54
|
Younger DS. Childhood muscular dystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:461-496. [PMID: 37562882 DOI: 10.1016/b978-0-323-98818-6.00024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Infancy- and childhood-onset muscular dystrophies are associated with a characteristic distribution and progression of motor dysfunction. The underlying causes of progressive childhood muscular dystrophies are heterogeneous involving diverse genetic pathways and genes that encode proteins of the plasma membrane, extracellular matrix, sarcomere, and nuclear membrane components. The prototypical clinicopathological features in an affected child may be adequate to fully distinguish it from other likely diagnoses based on four common features: (1) weakness and wasting of pelvic-femoral and scapular muscles with involvement of heart muscle; (2) elevation of serum muscle enzymes in particular serum creatine kinase; (3) necrosis and regeneration of myofibers; and (4) molecular neurogenetic assessment particularly utilizing next-generation sequencing of the genome of the likeliest candidates genes in an index case or family proband. A number of different animal models of therapeutic strategies have been developed for gene transfer therapy, but so far these techniques have not yet entered clinical practice. Treatment remains for the most part symptomatic with the goal of ameliorating locomotor and cardiorespiratory manifestations of the disease.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
55
|
Genetic Mapping of Behavioral Traits Using the Collaborative Cross Resource. Int J Mol Sci 2022; 24:ijms24010682. [PMID: 36614124 PMCID: PMC9821145 DOI: 10.3390/ijms24010682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
The complicated interactions between genetic background, environment and lifestyle factors make it difficult to study the genetic basis of complex phenotypes, such as cognition and anxiety levels, in humans. However, environmental and other factors can be tightly controlled in mouse studies. The Collaborative Cross (CC) is a mouse genetic reference population whose common genetic and phenotypic diversity is on par with that of humans. Therefore, we leveraged the power of the CC to assess 52 behavioral measures associated with locomotor activity, anxiety level, learning and memory. This is the first application of the CC in novel object recognition tests, Morris water maze tasks, and fear conditioning tests. We found substantial continuous behavioral variations across the CC strains tested, and mapped six quantitative trait loci (QTLs) which influenced these traits, defining candidate genetic variants underlying these QTLs. Overall, our findings highlight the potential of the CC population in behavioral genetic research, while the identified genomic loci and genes driving the variation of relevant behavioral traits provide a foundation for further studies.
Collapse
|
56
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
57
|
Lloyd EM, Pinniger GJ, Grounds MD, Murphy RM. Dysferlin Deficiency Results in Myofiber-Type Specific Differences in Abundances of Calcium-Handling and Glycogen Metabolism Proteins. Int J Mol Sci 2022; 24:ijms24010076. [PMID: 36613515 PMCID: PMC9820290 DOI: 10.3390/ijms24010076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by a genetic deficiency of the membrane-associated protein dysferlin, which usually manifest post-growth in young adults. The disease is characterized by progressive skeletal muscle wasting in the limb-girdle and limbs, inflammation, accumulation of lipid droplets in slow-twitch myofibers and, in later stages, replacement of muscles by adipose tissue. Previously we reported myofiber-type specific differences in muscle contractile function of 10-month-old dysferlin-deficient BLAJ mice that could not be fully accounted for by altered myofiber-type composition. In order to further investigate these findings, we examined the impact of dysferlin deficiency on the abundance of calcium (Ca2+) handling and glucose/glycogen metabolism-related proteins in predominantly slow-twitch, oxidative soleus and fast-twitch, glycolytic extensor digitorum longus (EDL) muscles of 10-month-old wild-type (WT) C57BL/6J and dysferlin-deficient BLAJ male mice. Additionally, we compared the Ca2+ activation properties of isolated slow- and fast-twitch myofibers from 3-month-old WT and BLAJ male mice. Differences were observed for some Ca2+ handling and glucose/glycogen metabolism-related protein levels between BLAJ soleus and EDL muscles (compared with WT) that may contribute to the previously reported differences in function in these BLAJ muscles. Dysferlin deficiency did not impact glycogen content of whole muscles nor Ca2+ activation of the myofilaments, although soleus muscle from 10-month-old BLAJ mice had more glycogen than EDL muscles. These results demonstrate a further impact of dysferlin deficiency on proteins associated with excitation-contraction coupling and glycogen metabolism in skeletal muscles, potentially contributing to altered contractile function in dysferlinopathy.
Collapse
Affiliation(s)
- Erin M. Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gavin J. Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Correspondence:
| | - Robyn M. Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
58
|
Pokrovsky MV, Korokin MV, Krayushkina AM, Zhunusov NS, Lapin KN, Soldatova MO, Kuzmin EA, Gudyrev OS, Kochkarova IS, Deikin AV. CONVENTIONAL APPROACHES TO THE THERAPY OF HEREDITARY MYOPATHIES. PHARMACY & PHARMACOLOGY 2022. [DOI: 10.19163/2307-9266-2022-10-5-416-431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the work was to analyze the available therapeutic options for the conventional therapy of hereditary myopathies.Materials and methods. When searching for the material for writing a review article, such abstract databases as PubMed and Google Scholar were used. The search was carried out on the publications during the period from 1980 to September 2022. The following words and their combinations were selected as parameters for the literature selection: “myopathy”, “Duchenne”, “myodystrophy”, “metabolic”, “mitochondrial”, “congenital”, “symptoms”, “replacement”, “recombinant”, “corticosteroids”, “vitamins”, “tirasemtiv”, “therapy”, “treatment”, “evidence”, “clinical trials”, “patients”, “dichloracetate”.Results. Congenital myopathies are a heterogeneous group of pathologies that are caused by atrophy and degeneration of muscle fibers due to mutations in genes. Based on a number of clinical and pathogenetic features, hereditary myopathies are divided into: 1) congenital myopathies; 2) muscular dystrophy; 3) mitochondrial and 4) metabolic myopathies. At the same time, treatment approaches vary significantly depending on the type of myopathy and can be based on 1) substitution of the mutant protein; 2) an increase in its expression; 3) stimulation of the internal compensatory pathways expression; 4) restoration of the compounds balance associated with the mutant protein function (for enzymes); 5) impact on the mitochondrial function (with metabolic and mitochondrial myopathies); 6) reduction of inflammation and fibrosis (with muscular dystrophies); as well as 7) an increase in muscle mass and strength. The current review presents current data on each of the listed approaches, as well as specific pharmacological agents with a description of their action mechanisms.Conclusion. Currently, the following pharmacological groups are used or undergoing clinical trials for the treatment of various myopathies types: inotropic, anti-inflammatory and antifibrotic drugs, antimyostatin therapy and the drugs that promote translation through stop codons (applicable for nonsense mutations). In addition, metabolic drugs, metabolic enzyme cofactors, mitochondrial biogenesis stimulators, and antioxidants can be used to treat myopathies. Finally, the recombinant drugs alglucosidase and avalglucosidase have been clinically approved for the replacement therapy of metabolic myopathies (Pompe’s disease).
Collapse
Affiliation(s)
| | | | | | | | - K. N. Lapin
- V.A. Negovsky Research Institute of General Reanimatology, Federal Scientific and Clinical Center for Resuscitation and Rehabilitology
| | | | - E. A. Kuzmin
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | |
Collapse
|
59
|
Inherited myopathies in the Middle East and North Africa. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
60
|
Tran V, Nahlé S, Robert A, Desanlis I, Killoran R, Ehresmann S, Thibault MP, Barford D, Ravichandran KS, Sauvageau M, Smith MJ, Kmita M, Côté JF. Biasing the conformation of ELMO2 reveals that myoblast fusion can be exploited to improve muscle regeneration. Nat Commun 2022; 13:7077. [PMID: 36400788 PMCID: PMC9674853 DOI: 10.1038/s41467-022-34806-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Myoblast fusion is fundamental for the development of multinucleated myofibers. Evolutionarily conserved proteins required for myoblast fusion include RAC1 and its activator DOCK1. In the current study we analyzed the contribution of the DOCK1-interacting ELMO scaffold proteins to myoblast fusion. When Elmo1-/- mice underwent muscle-specific Elmo2 genetic ablation, they exhibited severe myoblast fusion defects. A mutation in the Elmo2 gene that reduced signaling resulted in a decrease in myoblast fusion. Conversely, a mutation in Elmo2 coding for a protein with an open conformation increased myoblast fusion during development and in muscle regeneration. Finally, we showed that the dystrophic features of the Dysferlin-null mice, a model of limb-girdle muscular dystrophy type 2B, were reversed when expressing ELMO2 in an open conformation. These data provide direct evidence that the myoblast fusion process could be exploited for regenerative purposes and improve the outcome of muscle diseases.
Collapse
Affiliation(s)
- Viviane Tran
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Sarah Nahlé
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Amélie Robert
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
| | - Inès Desanlis
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Ryan Killoran
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Sophie Ehresmann
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | | | - David Barford
- MRC Laboratory of Molecular Biology, Cambridge, CB2 OQH, UK
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, 22908, VA, USA
- VIB/UGent Inflammation Research Centre, Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Martin Sauvageau
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Department of Biochemistry, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Matthew J Smith
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3T 1J4, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Marie Kmita
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Experimental Medicine, McGill University, Montréal, QC, H3G 2M1, Canada
| | - Jean-François Côté
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada.
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 0C7, Canada.
| |
Collapse
|
61
|
Winter L, Kustermann M, Ernhofer B, Höger H, Bittner RE, Schmidt WM. Proteins implicated in muscular dystrophy and cancer are functional constituents of the centrosome. Life Sci Alliance 2022; 5:e202201367. [PMID: 35790299 PMCID: PMC9259872 DOI: 10.26508/lsa.202201367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 12/02/2022] Open
Abstract
Aberrant expression of dystrophin, utrophin, dysferlin, or calpain-3 was originally identified in muscular dystrophies (MDs). Increasing evidence now indicates that these proteins might act as tumor suppressors in myogenic and non-myogenic cancers. As DNA damage and somatic aneuploidy, hallmarks of cancer, are early pathological signs in MDs, we hypothesized that a common pathway might involve the centrosome. Here, we show that dystrophin, utrophin, dysferlin, and calpain-3 are functional constituents of the centrosome. In myoblasts, lack of any of these proteins caused excess centrosomes, centrosome misorientation, nuclear abnormalities, and impaired microtubule nucleation. In dystrophin double-mutants, these defects were significantly aggravated. Moreover, we demonstrate that also in non-myogenic cells, all four MD-related proteins localize to the centrosome, including the muscle-specific full-length dystrophin isoform. Therefore, MD-related proteins might share a convergent function at the centrosome in addition to their diverse, well-established muscle-specific functions. Thus, our findings support the notion that cancer-like centrosome-related defects underlie MDs and establish a novel concept linking MDs to cancer.
Collapse
Affiliation(s)
- Lilli Winter
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Monika Kustermann
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Büsra Ernhofer
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Harald Höger
- Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Himberg, Austria
| | - Reginald E Bittner
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang M Schmidt
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
62
|
Devkota S, Shrestha S, Gurung T, Shrestha S. Regional anesthesia as a safe option in patient with limb girdle muscular dystrophy undergoing total abdominal hysterectomy: A case report and case review. Clin Case Rep 2022; 10:e6523. [PMID: 36381058 PMCID: PMC9637942 DOI: 10.1002/ccr3.6523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/25/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Regional anesthesia can be a very safe option in patients with limb girdle muscular dystrophy undergoing lower abdominal surgeries as general anesthesia and volatile anesthetic agents are associated with increased risk of malignant hyperthermia and rhabdomyolysis.
Collapse
Affiliation(s)
- Sagar Devkota
- Department of Anesthesiology and Critical CareSindhuli HospitalSindhuliNepal
| | | | - Tara Gurung
- Department of AnesthesiologyPMWHKathmanduNepal
| | - Saurav Shrestha
- Department of Anesthesiology and Critical CareNepal APF HospitalKathmanduNepal
| |
Collapse
|
63
|
Wang N, Han X, Hao S, Han J, Zhou X, Sun S, Tang J, Lu Y, Wu H, Ma S, Song X, Ji G. The clinical, myopathological, and molecular characteristics of 26 Chinese patients with dysferlinopathy: a high proportion of misdiagnosis and novel variants. BMC Neurol 2022; 22:398. [PMID: 36319958 PMCID: PMC9623978 DOI: 10.1186/s12883-022-02905-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Dysferlinopathy is an autosomal recessive muscular dystrophy caused by pathogenic variants in the dysferlin (DYSF) gene. This disease shows heterogeneous clinical phenotypes and genetic characteristics. METHODS We reviewed the clinical and pathological data as well as the molecular characteristics of 26 Chinese patients with dysferlinopathy screened by immunohistochemistry staining and pathogenic variants in DYSF genes. RESULTS Among 26 patients with dysferlinopathy, 18 patients (69.2%) presented as Limb-girdle Muscular Dystrophy Type R2 (LGMD R2), 4 (15.4%) had a phenotype of Miyoshi myopathy (MM), and 4 (15.4%) presented as asymptomatic hyperCKemia. Fifteen patients (57.7%) were originally misdiagnosed as inflammatory myopathy or other diseases. Fifteen novel variants were identified among the 40 variant sites identified in this cohort. CONCLUSION Dysferlinopathy is a clinically and genetically heterogeneous group of disorders with various phenotypes, a high proportion of novel variants, and a high rate of misdiagnosis before immunohistochemistry staining and genetic analysis.
Collapse
Affiliation(s)
- Ning Wang
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Xu Han
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Shengpu Hao
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Jingzhe Han
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | | | - Shuyan Sun
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Jin Tang
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Yanpeng Lu
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Hongran Wu
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Shaojuan Ma
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Xueqin Song
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Guang Ji
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| |
Collapse
|
64
|
Xie Y, Li YH, Chen K, Zhu CY, Bai JY, Xiao F, Tan S, Zeng L. Key biomarkers and latent pathways of dysferlinopathy: Bioinformatics analysis and in vivo validation. Front Neurol 2022; 13:998251. [PMID: 36203997 PMCID: PMC9530905 DOI: 10.3389/fneur.2022.998251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background Dysferlinopathy refers to a group of muscle diseases with progressive muscle weakness and atrophy caused by pathogenic mutations of the DYSF gene. The pathogenesis remains unknown, and currently no specific treatment is available to alter the disease progression. This research aims to investigate important biomarkers and their latent biological pathways participating in dysferlinopathy and reveal the association with immune cell infiltration. Methods GSE3307 and GSE109178 were obtained from the Gene Expression Omnibus (GEO) database. Based on weighted gene co-expression network analysis (WGCNA) and differential expression analysis, coupled with least absolute shrinkage and selection operator (LASSO), the key genes for dysferlinopathy were identified. Functional enrichment analysis Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were applied to disclose the hidden biological pathways. Following that, the key genes were approved for diagnostic accuracy of dysferlinopathy based on another dataset GSE109178, and quantitative real-time polymerase chain reaction (qRT-PCR) were executed to confirm their expression. Furthermore, the 28 immune cell abundance patterns in dysferlinopathy were determined with single-sample GSEA (ssGSEA). Results 1,579 differentially expressed genes (DEGs) were screened out. Based on WGCNA, three co-expression modules were obtained, with the MEskyblue module most strongly correlated with dysferlinopathy. 44 intersecting genes were recognized from the DEGs and the MEskyblue module. The six key genes MVP, GRN, ERP29, RNF128, NFYB and KPNA3 were discovered through LASSO analysis and experimentally verified later. In a receiver operating characteristic analysis (ROC) curve, the six hub genes were shown to be highly valuable for diagnostic purposes. Furthermore, functional enrichment analysis highlighted that these genes were enriched mainly along the ubiquitin-proteasome pathway (UPP). Ultimately, ssGSEA showed a significant immune-cell infiltrative microenvironment in dysferlinopathy patients, especially T cell, macrophage, and activated dendritic cell (DC). Conclusion Six key genes are identified in dysferlinopathy with a bioinformatic approach used for the first time. The key genes are believed to be involved in protein degradation pathways and the activation of muscular inflammation. And several immune cells, such as T cell, macrophage and DC, are considered to be implicated in the progression of dysferlinopathy.
Collapse
Affiliation(s)
- Yan Xie
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ying-hui Li
- Department of Neurology, People's Hospital of Yilong County, Nanchong, China
| | - Kai Chen
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Chun-yan Zhu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jia-ying Bai
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Feng Xiao
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Song Tan
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Li Zeng
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- *Correspondence: Li Zeng
| |
Collapse
|
65
|
Hui J, Stjepić V, Nakamura M, Parkhurst SM. Wrangling Actin Assemblies: Actin Ring Dynamics during Cell Wound Repair. Cells 2022; 11:2777. [PMID: 36139352 PMCID: PMC9497110 DOI: 10.3390/cells11182777] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 12/18/2022] Open
Abstract
To cope with continuous physiological and environmental stresses, cells of all sizes require an effective wound repair process to seal breaches to their cortex. Once a wound is recognized, the cell must rapidly plug the injury site, reorganize the cytoskeleton and the membrane to pull the wound closed, and finally remodel the cortex to return to homeostasis. Complementary studies using various model organisms have demonstrated the importance and complexity behind the formation and translocation of an actin ring at the wound periphery during the repair process. Proteins such as actin nucleators, actin bundling factors, actin-plasma membrane anchors, and disassembly factors are needed to regulate actin ring dynamics spatially and temporally. Notably, Rho family GTPases have been implicated throughout the repair process, whereas other proteins are required during specific phases. Interestingly, although different models share a similar set of recruited proteins, the way in which they use them to pull the wound closed can differ. Here, we describe what is currently known about the formation, translocation, and remodeling of the actin ring during the cell wound repair process in model organisms, as well as the overall impact of cell wound repair on daily events and its importance to our understanding of certain diseases and the development of therapeutic delivery modalities.
Collapse
Affiliation(s)
| | | | | | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
66
|
Ivanova A, Smirnikhina S, Lavrov A. Dysferlinopathies: clinical and genetic variability. Clin Genet 2022; 102:465-473. [PMID: 36029111 DOI: 10.1111/cge.14216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022]
Abstract
Dysferlinopathies are a clinically heterogeneous group of diseases caused by mutations in the DYSF gene encoding the dysferlin protein. Dysferlin is mostly expressed in muscle tissues and is localized in the sarcolemma, where it performs its main function of resealing and maintaining of the integrity of the cell membrane. At least four forms of dysferlinopathies have been described: Miyoshi myopathy, limb-girdle muscular dystrophy type 2B, distal myopathy with anterior tibial onset, and isolated hyperCKemia. Here we review the clinical features of different forms of dysferlinopathies and attempt to identify genotype-phenotype correlations. Because of the great clinical variability and rarety of the disease and mutations little is known, how different phenotypes develop as a result of different mutations. However missense mutations seem to induce more severe disease than LoF, which is typical for many muscle dystrophies. The role of several specific mutations and possible gene modifiers is also discussed in the paper.
Collapse
Affiliation(s)
- Alisa Ivanova
- Research Centre for Medical Genetics, Moskvorechye 1, Moscow, Russia
| | | | - Alexander Lavrov
- Research Centre for Medical Genetics, Moskvorechye 1, Moscow, Russia
| |
Collapse
|
67
|
Folland C, Johnsen R, Gomez AB, Trajanoski D, Davis MR, Moore U, Straub V, Barresi R, Guglieri M, Hayhurst H, Schaefer AM, Laing NG, Lamont PJ, Ravenscroft G. Identification of a novel heterozygous DYSF variant in a large family with a dominantly-inherited dysferlinopathy. Neuropathol Appl Neurobiol 2022; 48:e12846. [PMID: 35962550 DOI: 10.1111/nan.12846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/29/2022] [Accepted: 08/07/2022] [Indexed: 11/27/2022]
Abstract
AIMS Dysferlinopathy is an autosomal recessive muscular dystrophy, caused by bi-allelic variants in the gene encoding dysferlin (DYSF). Onset typically occurs in the second to third decade and is characterised by slowly progressive skeletal muscle weakness and atrophy of the proximal and/or distal muscles of the four limbs. There are rare cases of symptomatic DYSF variant carriers. Here, we report a large family with a dominantly inherited hyperCKaemia and late-onset muscular dystrophy. METHODS AND RESULTS Genetic analysis identified a co-segregating novel DYSF variant [NM_003494.4:c.6207del p.(Tyr2070Metfs*4)]. No secondary variants in DYSF or other dystrophy-related genes were identified on whole genome sequencing and analysis of the proband's DNA. Skeletal muscle involvement was milder and later onset than typical dysferlinopathy presentations; these clinical signs manifested in four individuals, all between the fourth and sixth decades of life. All individuals heterozygous for the c.6207del variant had hyperCKaemia. Histological analysis of skeletal muscle biopsies across three generations showed clear dystrophic signs, including inflammatory infiltrates, regenerating myofibres, increased variability in myofibre size, and internal nuclei. Muscle magnetic resonance imaging revealed fatty replacement of muscle in two individuals. Western blot and immunohistochemical analysis of muscle biopsy demonstrated consistent reduction of dysferlin staining. Allele-specific quantitative PCR analysis of DYSF mRNA from patient muscle found that the variant, localised to the extreme C-terminus of dysferlin, does not activate post-transcriptional mRNA decay. CONCLUSIONS We propose that this inheritance pattern may be underappreciated and that other late-onset muscular dystrophy cases with mono-allelic DYSF variants, particularly C-terminal premature truncation variants, may represent dominant forms of disease.
Collapse
Affiliation(s)
- Chiara Folland
- Centre for Medical Research, University of Western Australia, Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | - Russell Johnsen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, Australia
| | - Adriana Botero Gomez
- Department of Diagnostic Genomics, Department of Health, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Daniel Trajanoski
- Department of Diagnostic Genomics, Department of Health, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Mark R Davis
- Department of Diagnostic Genomics, Department of Health, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Ursula Moore
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Hannah Hayhurst
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Andrew M Schaefer
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Nigel G Laing
- Centre for Medical Research, University of Western Australia, Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | | | - Gianina Ravenscroft
- Centre for Medical Research, University of Western Australia, Harry Perkins Institute of Medical Research, Perth, WA, Australia
| |
Collapse
|
68
|
A Novel Homozygous Variant in DYSF Gene Is Associated with Autosomal Recessive Limb Girdle Muscular Dystrophy R2/2B. Int J Mol Sci 2022; 23:ijms23168932. [PMID: 36012197 PMCID: PMC9408934 DOI: 10.3390/ijms23168932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in the DYSF gene, encoding dysferlin, are responsible for Limb Girdle Muscular Dystrophy type R2/2B (LGMDR2/2B), Miyoshi myopathy (MM), and Distal Myopathy with Anterior Tibialis onset (MDAT). The size of the gene and the reported inter and intra familial phenotypic variability make early diagnosis difficult. Genetic analysis was conducted using Next Gene Sequencing (NGS), with a panel of 40 Muscular Dystrophies associated genes we designed. In the present study, we report a new missense variant c.5033G>A, p.Cys1678Tyr (NM_003494) in the exon 45 of DYSF gene related to Limb Girdle Muscular Dystrophy type R2/2B in a 57-year-old patient affected with LGMD from a consanguineous family of south Italy. Both healthy parents carried this variant in heterozygosity. Genetic analysis extended to two moderately affected sisters of the proband, showed the presence of the variant c.5033G>A in both in homozygosity. These data indicate a probable pathological role of the variant c.5033G>A never reported before in the onset of LGMDR2/2B, pointing at the NGS as powerful tool for identifying LGMD subtypes. Moreover, the collection and the networking of genetic data will increase power of genetic-molecular investigation, the management of at-risk individuals, the development of new therapeutic targets and a personalized medicine.
Collapse
|
69
|
Dalakas MC, Dalmau J. Comment: Humoral and T-cell Immunities to SARS-CoV-2 Vaccines: Safety, Efficacy, and Challenges in Autoimmune Neurology. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:e200010. [PMID: 35728948 PMCID: PMC9219497 DOI: 10.1212/nxi.0000000000200010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Marinos C Dalakas
- From the Thomas Jefferson University (M.C.D.), Philadelphia, PA; University of Athens Medical School (M.C.D.), Greece; IDIBAPS-Hospital Clinic of Barcelona (J.D.), University of Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, PA; and Catalan Institution for Research and Advanced Studies (ICREA) (J.D.), Barcelona, Spain.
| | - Josep Dalmau
- From the Thomas Jefferson University (M.C.D.), Philadelphia, PA; University of Athens Medical School (M.C.D.), Greece; IDIBAPS-Hospital Clinic of Barcelona (J.D.), University of Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, PA; and Catalan Institution for Research and Advanced Studies (ICREA) (J.D.), Barcelona, Spain.
| |
Collapse
|
70
|
Singhal R, Lukose R, Carr G, Moktar A, Gonzales-Urday AL, Rouchka EC, Vajravelu BN. Differential Expression of Long Noncoding RNAs in Murine Myoblasts After Short Hairpin RNA-Mediated Dysferlin Silencing In Vitro: Microarray Profiling. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2022; 3:e33186. [PMID: 38935964 PMCID: PMC11135227 DOI: 10.2196/33186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/02/2022] [Accepted: 05/10/2022] [Indexed: 06/29/2024]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are noncoding RNA transcripts greater than 200 nucleotides in length and are known to play a role in regulating the transcription of genes involved in vital cellular functions. We hypothesized the disease process in dysferlinopathy is linked to an aberrant expression of lncRNAs and messenger RNAs (mRNAs). OBJECTIVE In this study, we compared the lncRNA and mRNA expression profiles between wild-type and dysferlin-deficient murine myoblasts (C2C12 cells). METHODS LncRNA and mRNA expression profiling were performed using a microarray. Several lncRNAs with differential expression were validated using quantitative real-time polymerase chain reaction. Gene Ontology (GO) analysis was performed to understand the functional role of the differentially expressed mRNAs. Further bioinformatics analysis was used to explore the potential function, lncRNA-mRNA correlation, and potential targets of the differentially expressed lncRNAs. RESULTS We found 3195 lncRNAs and 1966 mRNAs that were differentially expressed. The chromosomal distribution of the differentially expressed lncRNAs and mRNAs was unequal, with chromosome 2 having the highest number of lncRNAs and chromosome 7 having the highest number of mRNAs that were differentially expressed. Pathway analysis of the differentially expressed genes indicated the involvement of several signaling pathways including PI3K-Akt, Hippo, and pathways regulating the pluripotency of stem cells. The differentially expressed genes were also enriched for the GO terms, developmental process and muscle system process. Network analysis identified 8 statistically significant (P<.05) network objects from the upregulated lncRNAs and 3 statistically significant network objects from the downregulated lncRNAs. CONCLUSIONS Our results thus far imply that dysferlinopathy is associated with an aberrant expression of multiple lncRNAs, many of which may have a specific function in the disease process. GO terms and network analysis suggest a muscle-specific role for these lncRNAs. To elucidate the specific roles of these abnormally expressed noncoding RNAs, further studies engineering their expression are required.
Collapse
Affiliation(s)
- Richa Singhal
- Department of Biochemistry and Molecular Genetics, KY IDeA Networks of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY, United States
| | - Rachel Lukose
- Department of Physician Assistant Studies, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Gwenyth Carr
- Department of Medical and Molecular Biology, School of Arts and Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Afsoon Moktar
- Department of Physician Assistant Studies, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Ana Lucia Gonzales-Urday
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Eric C Rouchka
- Department of Biochemistry and Molecular Genetics, KY IDeA Networks of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY, United States
| | - Bathri N Vajravelu
- Department of Physician Assistant Studies, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| |
Collapse
|
71
|
Reyngoudt H, Smith FE, Caldas de Almeida Araújo E, Wilson I, Fernández-Torrón R, James MK, Moore UR, Díaz-Manera J, Marty B, Azzabou N, Gordish H, Rufibach L, Hodgson T, Wallace D, Ward L, Boisserie JM, Le Louër J, Hilsden H, Sutherland H, Canal A, Hogrel JY, Jacobs M, Stojkovic T, Bushby K, Mayhew A, Straub V, Carlier PG, Blamire AM. Three-year quantitative magnetic resonance imaging and phosphorus magnetic resonance spectroscopy study in lower limb muscle in dysferlinopathy. J Cachexia Sarcopenia Muscle 2022; 13:1850-1863. [PMID: 35373496 PMCID: PMC9178361 DOI: 10.1002/jcsm.12987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Natural history studies in neuromuscular disorders are vital to understand the disease evolution and to find sensitive outcome measures. We performed a longitudinal assessment of quantitative magnetic resonance imaging (MRI) and phosphorus magnetic resonance spectroscopy (31 P MRS) outcome measures and evaluated their relationship with function in lower limb skeletal muscle of dysferlinopathy patients. METHODS Quantitative MRI/31 P MRS data were obtained at 3 T in two different sites in 54 patients and 12 controls, at baseline, and three annual follow-up visits. Fat fraction (FF), contractile cross-sectional area (cCSA), and muscle water T2 in both global leg and thigh segments and individual muscles and 31 P MRS indices in the anterior leg compartment were assessed. Analysis included comparisons between patients and controls, assessments of annual changes using a linear mixed model, standardized response means (SRM), and correlations between MRI and 31 P MRS markers and functional markers. RESULTS Posterior muscles in thigh and leg showed the highest FF values. FF at baseline was highly heterogeneous across patients. In ambulant patients, median annual increases in global thigh and leg segment FF values were 4.1% and 3.0%, respectively (P < 0.001). After 3 years, global thigh and leg FF increases were 9.6% and 8.4%, respectively (P < 0.001). SRM values for global thigh FF were over 0.8 for all years. Vastus lateralis muscle showed the highest SRM values across all time points. cCSA decreased significantly after 3 years with median values of 11.0% and 12.8% in global thigh and global leg, respectively (P < 0.001). Water T2 values in ambulant patients were significantly increased, as compared with control values (P < 0.001). The highest water T2 values were found in the anterior part of thigh and leg. Almost all 31 P MRS indices were significantly different in patients as compared with controls (P < 0.006), except for pHw , and remained, similar as to water T2 , abnormal for the whole study duration. Global thigh water T2 at baseline was significantly correlated to the change in FF after 3 years (ρ = 0.52, P < 0.001). There was also a significant relationship between the change in functional score and change in FF after 3 years in ambulant patients (ρ = -0.55, P = 0.010). CONCLUSIONS This multi-centre study has shown that quantitative MRI/31 P MRS measurements in a heterogeneous group of dysferlinopathy patients can measure significant changes over the course of 3 years. These data can be used as reference values in view of future clinical trials in dysferlinopathy or comparisons with quantitative MRI/S data obtained in other limb-girdle muscular dystrophy subtypes.
Collapse
Affiliation(s)
- Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Fiona E Smith
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ericky Caldas de Almeida Araújo
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Ian Wilson
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Roberto Fernández-Torrón
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Neuromuscular Area, Biodonostia Health Research Institute, Neurology Service, Donostia University Hospital, Donostia-San Sebastian, Spain
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ursula R Moore
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jordi Díaz-Manera
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Neuromuscular Disorders Unit, Neurology Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain
| | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Noura Azzabou
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Heather Gordish
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, USA.,Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, DC, USA
| | | | - Tim Hodgson
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dorothy Wallace
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Louise Ward
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jean-Marc Boisserie
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Julien Le Louër
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Heather Hilsden
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Helen Sutherland
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Aurélie Canal
- Neuromuscular Physiology and Evaluation Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Jean-Yves Hogrel
- Neuromuscular Physiology and Evaluation Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Marni Jacobs
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, USA.,Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, DC, USA
| | - Tanya Stojkovic
- Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital (AP-HP), Paris, France
| | - Kate Bushby
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Andrew M Blamire
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
72
|
Reash NF, James MK, Alfano LN, Mayhew AG, Jacobs M, Iammarino MA, Holsten S, Sakamoto C, Tateishi T, Yajima H, Duong T, de Wolf B, Gee R, Bharucha-Goebel DX, Bravver E, Mori-Yoshimura M, Bushby K, Rufibach LE, Straub V, Lowes LP. Comparison of strength testing modalities in dysferlinopathy. Muscle Nerve 2022; 66:159-166. [PMID: 35506767 DOI: 10.1002/mus.27570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION/AIMS Dysferlinopathy demonstrates heterogeneity in muscle weakness between patients, which can progress at different rates over time. Changing muscle strength due to disease progression or from an investigational product is associated with changing functional ability. The purpose of this study was to compare three methods of strength testing used in the Clinical Outcome Study (COS) for dysferlinopathy to understand which method and which muscle groups were most sensitive to change over time. METHODS Patients were evaluated at each study visit using functional scales, manual muscle testing, and handheld dynamometry (HHD) at all 15 sites. A fixed-frame system (Fixed) was used at a subset of seven sites. Screening and baseline visits were evaluated for reliability. Data over a 1-year period were analyzed to determine sensitivity to change among strength modalities and individual muscle groups. RESULTS HHD and Fixed captured significant change across 1 year in summed muscle strength score of four muscle groups (P < .01). Strength summed scores were significantly correlated with functional scales (rho = 0.68-0.92, P < .001). Individual muscle groups, however, showed high levels of variability between visits. DISCUSSION Although both HHD and Fixed demonstrate change over 12 months, HHD is a less expensive option that provides data on a continuous scale and may be easier to implement. Due to variability in strength measures, researchers should carefully consider use of strength testing as an outcome and may wish to select functional measures with less variability as clinical trial endpoints.
Collapse
Affiliation(s)
- Natalie F Reash
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Lindsay N Alfano
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Anna G Mayhew
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Marni Jacobs
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC.,Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, DC
| | - Megan A Iammarino
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Scott Holsten
- Neuroscience Institute, Carolinas Neuromuscular/ALS-MDA Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Chikako Sakamoto
- Department of Physical Rehabilitation, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takayuki Tateishi
- Department of Physical Rehabilitation, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroyuki Yajima
- Department of Physical Rehabilitation, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tina Duong
- Cooperative International Neuromuscular Research Group, Children's National Health System, Washington, DC.,Lucile Salter Packard Children's Hospital at Stanford, Neurology, Palo Alto, California.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Brittney de Wolf
- Cooperative International Neuromuscular Research Group, Children's National Health System, Washington, DC
| | - Richard Gee
- Lucile Salter Packard Children's Hospital at Stanford, Neurology, Palo Alto, California
| | - Diana X Bharucha-Goebel
- Department of Neurology Children's National Health System, Washington, DC.,National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Elena Bravver
- Neuroscience Institute, Carolinas Neuromuscular/ALS-MDA Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kate Bushby
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Linda P Lowes
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | | |
Collapse
|
73
|
Advanced Gene-Targeting Therapies for Motor Neuron Diseases and Muscular Dystrophies. Int J Mol Sci 2022; 23:ijms23094824. [PMID: 35563214 PMCID: PMC9101723 DOI: 10.3390/ijms23094824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/19/2022] Open
Abstract
Gene therapy is a revolutionary, cutting-edge approach to permanently ameliorate or amend many neuromuscular diseases by targeting their genetic origins. Motor neuron diseases and muscular dystrophies, whose genetic causes are well known, are the frontiers of this research revolution. Several genetic treatments, with diverse mechanisms of action and delivery methods, have been approved during the past decade and have demonstrated remarkable results. However, despite the high number of genetic treatments studied preclinically, those that have been advanced to clinical trials are significantly fewer. The most clinically advanced treatments include adeno-associated virus gene replacement therapy, antisense oligonucleotides, and RNA interference. This review provides a comprehensive overview of the advanced gene therapies for motor neuron diseases (i.e., amyotrophic lateral sclerosis and spinal muscular atrophy) and muscular dystrophies (i.e., Duchenne muscular dystrophy, limb-girdle muscular dystrophy, and myotonic dystrophy) tested in clinical trials. Emphasis has been placed on those methods that are a few steps away from their authoritative approval.
Collapse
|
74
|
Paleo BJ, McElhanon KE, Bulgart HR, Banford KK, Beck EX, Sattler KM, Goines BN, Ratcliff SL, Crowe KE, Weisleder N. Reduced Sarcolemmal Membrane Repair Exacerbates Striated Muscle Pathology in a Mouse Model of Duchenne Muscular Dystrophy. Cells 2022; 11:1417. [PMID: 35563723 PMCID: PMC9100510 DOI: 10.3390/cells11091417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a common X-linked degenerative muscle disorder that involves mutations in the DMD gene that frequently reduce the expression of the dystrophin protein, compromising the structural integrity of the sarcolemmal membrane and leaving it vulnerable to injury during cycles of muscle contraction and relaxation. This results in an increased frequency of sarcolemma disruptions that can compromise the barrier function of the membrane and lead to death of the myocyte. Sarcolemmal membrane repair processes can potentially compensate for increased membrane disruptions in DMD myocytes. Previous studies demonstrated that TRIM72, a muscle-enriched tripartite motif (TRIM) family protein also known as mitsugumin 53 (MG53), is a component of the cell membrane repair machinery in striated muscle. To test the importance of membrane repair in striated muscle in compensating for the membrane fragility in DMD, we crossed TRIM72/MG53 knockout mice into the mdx mouse model of DMD. These double knockout (DKO) mice showed compromised sarcolemmal membrane integrity compared to mdx mice, as measured by immunoglobulin G staining and ex vivo muscle laser microscopy wounding assays. We also found a significant decrease in muscle ex vivo contractile function as compared to mdx mice at both 6 weeks and 1.5 years of age. As the DKO mice aged, they developed more extensive fibrosis in skeletal muscles compared to mdx. Our findings indicate that TRIM72/MG53-mediated membrane repair can partially compensate for the sarcolemmal fragility associated with DMD and that the loss of membrane repair results in increased pathology in the DKO mice.
Collapse
Affiliation(s)
- Brian J. Paleo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Kevin E. McElhanon
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Hannah R. Bulgart
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Kassidy K. Banford
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Eric X Beck
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Kristina M. Sattler
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Briana N. Goines
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Shelby L. Ratcliff
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Kelly E. Crowe
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| |
Collapse
|
75
|
Rossi D, Pierantozzi E, Amadsun DO, Buonocore S, Rubino EM, Sorrentino V. The Sarcoplasmic Reticulum of Skeletal Muscle Cells: A Labyrinth of Membrane Contact Sites. Biomolecules 2022; 12:488. [PMID: 35454077 PMCID: PMC9026860 DOI: 10.3390/biom12040488] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/17/2022] Open
Abstract
The sarcoplasmic reticulum of skeletal muscle cells is a highly ordered structure consisting of an intricate network of tubules and cisternae specialized for regulating Ca2+ homeostasis in the context of muscle contraction. The sarcoplasmic reticulum contains several proteins, some of which support Ca2+ storage and release, while others regulate the formation and maintenance of this highly convoluted organelle and mediate the interaction with other components of the muscle fiber. In this review, some of the main issues concerning the biology of the sarcoplasmic reticulum will be described and discussed; particular attention will be addressed to the structure and function of the two domains of the sarcoplasmic reticulum supporting the excitation-contraction coupling and Ca2+-uptake mechanisms.
Collapse
Affiliation(s)
- Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (E.P.); (D.O.A.); (S.B.); (E.M.R.); (V.S.)
| | | | | | | | | | | |
Collapse
|
76
|
Kraya T, Mensch A, Zierz S, Stoevesandt D, Nägel S. Update Distale Myopathien. KLIN NEUROPHYSIOL 2022. [DOI: 10.1055/a-1737-8273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ZusammenfassungDie Distalen Myopathien umfassen eine Gruppe von genetisch determinierten
Muskelerkrankungen bei denen Paresen und eine fortschreitende Atrophie der
distalen Muskelgruppen im Vordergrund stehen. Der klinische Phänotyp,
der Erkrankungsbeginn, der Vererbungsmodus sowie histologische
Veränderungen helfen die einzelnen Formen zu differenzieren. Das
klinische und genetische Spektrum ist allerdings heterogen. In den letzten
Jahren hat durch die erweiterte genetische Diagnostik die Anzahl der
nachgewiesenen Mutationen exponentiell zugenommen. Im folgenden Beitrag werden
die Klassifikation, die klinischen Besonderheiten und die relevanten genetischen
Aspekte dargestellt.
Collapse
Affiliation(s)
- Torsten Kraya
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
- Klinik für Neurologie, Klinikum St. Georg Leipzig
gGmbH
| | - Alexander Mensch
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Stephan Zierz
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Dietrich Stoevesandt
- Universitätsklinik und Poliklinik für Radiologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Steffen Nägel
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| |
Collapse
|
77
|
Alharbi N, Matar R, Cupler E, Al-Hindi H, Murad H, Alhomud I, Monies D, Alshehri A, Alyahya M, Meyer B, Bohlega S. Clinical, Neurophysiological, Radiological, Pathological, and Genetic Features of Dysferlinopathy in Saudi Arabia. Front Neurosci 2022; 16:815556. [PMID: 35273475 PMCID: PMC8902167 DOI: 10.3389/fnins.2022.815556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTo characterize the phenotypic, neurophysiological, radiological, pathological, and genetic profile of 33 Saudi Arabian families with dysferlinopathy.MethodsA descriptive observational study was done on a cohort of 112 Saudi Arabian families with LGMD. Screening for the Dysferlin (DYSF) gene was done in a tertiary care referral hospital in Saudi Arabia. Clinical, Neurophysiological, Radiological, Pathological, and Genetic findings in subjects with dysferlin mutation were the primary outcome variables. Statistical analysis was done by Epi-info.Results33 out of 112 families (29.46%) registered in the LGMD cohort had Dysferlinopathy. 53 subjects (28 males, 52.83%) from 33 families were followed up for various periods ranging from 1 to 28 years. The mean age of onset was 17.79 ± 3.48 years (Range 10 to 25 years). Miyoshi Myopathy phenotype was observed in 50.94% (27 out of 53), LGMDR2 phenotype in 30.19% (16 out of 53), and proximodistal phenotype in 15.09% (8 out of 53) of the subjects. Loss of ambulation was observed in 39.62% (21 out of 53 subjects). Electrophysiological, Radiological, and histopathological changes were compatible with the diagnosis. Mean serum Creatinine Kinase was 6,464.45 ± 4,149.24 with a range from 302 to 21,483 IU/L. In addition, 13 dysferlin mutations were identified two of them were compound heterozygous. One founder mutation was observed c.164_165insA in 19 unrelated families.ConclusionThe prevalence of Dysferlinopathy was 29.46% in the native Saudi LGMD cohort. It is the most prevalent subtype seconded by calpainopathy. The clinical course varied among the study subjects and was consistent with those reported from different ethnic groups. One founder mutation was identified. Initial screening of the founder mutations in new families is highly recommended.
Collapse
Affiliation(s)
- Norah Alharbi
- Department of Clinical Science, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Edward Cupler
- Department of Neuroscience, King Faisal Specialist Hospital, and Research Center, Jeddah, Saudi Arabia
| | - Hindi Al-Hindi
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hatem Murad
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Iftteah Alhomud
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Dorota Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ali Alshehri
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mossaed Alyahya
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Brian Meyer
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Saeed Bohlega
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- *Correspondence: Saeed Bohlega,
| |
Collapse
|
78
|
White Z, Theret M, Milad N, Tung LW, Chen WWH, Sirois MG, Rossi F, Bernatchez P. Cholesterol absorption blocker ezetimibe prevents muscle wasting in severe dysferlin-deficient and mdx mice. J Cachexia Sarcopenia Muscle 2022; 13:544-560. [PMID: 34927367 PMCID: PMC8818667 DOI: 10.1002/jcsm.12879] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Muscular dystrophy (MD) causes muscle wasting and is often lethal in patients due to a lack of proven therapies. In contrast, mouse models of MD are notoriously mild. We have previously shown severe human-like muscle pathology in mdx [Duchenne MD (DMD)] and dysferlin-deficient limb-girdle MD type 2B (LGMD2B) mice by inactivating the gene encoding for apolipoprotein E (ApoE), a lipid transporter synthesized by the liver, brain and adipocytes to regulate lipid and fat metabolism. Having recently established that human DMD is a novel type of primary genetic dyslipidaemia with elevated cholesterol, we sought to determine whether cholesterol could exacerbate the muscle wasting process observed in severe rodent MD. METHODS Severe mdx and dysferlin knock-out mice lacking ApoE were treated with ezetimibe (15 mg/kg/day), a clinically approved drug exhibiting few pleiotropic effects. In separate studies, dietary cholesterol was raised (from 0.2% to 2% cholesterol) in combination with experimental micro-injury and direct cholesterol injection assays. Muscles were assessed histologically for changes in collagen and adipocyte infiltration and both transcriptomic and cellular changes by RNA-seq and fluorescence-activated cell sorting analysis. RESULTS Treatment of severe DMD and LGMD2B mice with ezetimibe completely prevented clinical signs of ambulatory dysfunction (0% incidence vs. 33% for vehicle treatment; P < 0.05). Histological analyses revealed that ezetimibe-reduced fibro-fatty infiltration up to 84% and 63% in severely affected triceps (P ≤ 0.0001) and gastrocnemius (P ≤ 0.003) muscles, resulting in a respective 1.9-fold and 2.2-fold retention of healthy myofibre area (P ≤ 0.0001). Additionally, raising dietary cholesterol and thus concentrations of plasma low-density lipoprotein-associated cholesterol (by 250%; P < 0.0001) reduced overall survivability (by 100%; P < 0.001) and worsened muscle damage in the LGMD2B triceps by 767% (P < 0.03). Micro-pin-induced mechanical injury in LGMD2B mice fed a high cholesterol diet exacerbated muscle damage by 425% (P < 0.03) and increased macrophage recruitment (by 98%; P = 0.03) compared with those injured on a chow diet. Parallel RNA-seq analyses revealed that injury in cholesterol-fed mice also modulated the expression of 3671 transcripts (1953 up-regulated), with fibrogenic, inflammatory and programmed cell death-associated pathways among the most enriched. Mice lacking dysferlin also displayed heightened muscle necrosis (by 123%; P < 0.0001) following a direct intramuscular injection of cholesterol compared with control mice. CONCLUSIONS Cholesterol exacerbates rodent MD. Specific inhibition of cholesterol absorption with ezetimibe may safely attenuate human MD severity and delay death.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - William Wei-Han Chen
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Martin G Sirois
- Montreal Heart Institute, Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Fabio Rossi
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
79
|
Tominaga K, Tominaga N, Williams EO, Rufibach L, Schöwel V, Spuler S, Viswanathan M, Guarente LP. 4-Phenylbutyrate restores localization and membrane repair to human dysferlin mutations. iScience 2022; 25:103667. [PMID: 35028538 PMCID: PMC8741482 DOI: 10.1016/j.isci.2021.103667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 12/15/2021] [Indexed: 10/29/2022] Open
Abstract
Dysferlinopathies are muscular dystrophies caused by recessive loss-of-function mutations in dysferlin (DYSF), a membrane protein involved in skeletal muscle membrane repair. We describe a cell-based assay in which human DYSF proteins bearing missense mutations are quantitatively assayed for membrane localization by flow cytometry and identified 64 localization-defective DYSF mutations. Using this platform, we show that the clinically approved drug 4-phenylbutryric acid (4-PBA) partially restores membrane localization to 25 mutations, as well as membrane repair to cultured myotubes expressing 2 different mutations. Two-day oral administration of 4-PBA to mice homozygous for one of these mutations restored myofiber membrane repair. 4-PBA may hold therapeutic potential for treating a subset of humans with muscular dystrophy due to dysferlin deficiency.
Collapse
Affiliation(s)
- Kana Tominaga
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Pathology and Oncology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Naoomi Tominaga
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric O. Williams
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Fitchburg State University, School of Heath and Natural Sciences, Antonucci Science Complex 235, Fitchburg, MA 01420, USA
| | - Laura Rufibach
- Jain Foundation, 9706 4th Avenue NE, Suite 101, Seattle, WA 98115, USA
| | - Verena Schöwel
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation of Charité Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation of Charité Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mohan Viswanathan
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leonard P. Guarente
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
80
|
Bittel DC, Sreetama SC, Chandra G, Ziegler R, Nagaraju K, Van der Meulen JH, Jaiswal JK. Secreted acid sphingomyelinase as a potential gene therapy for limb girdle muscular dystrophy 2B. J Clin Invest 2022; 132:e141295. [PMID: 34981776 PMCID: PMC8718136 DOI: 10.1172/jci141295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Efficient sarcolemmal repair is required for muscle cell survival, with deficits in this process leading to muscle degeneration. Lack of the sarcolemmal protein dysferlin impairs sarcolemmal repair by reducing secretion of the enzyme acid sphingomyelinase (ASM), and causes limb girdle muscular dystrophy 2B (LGMD2B). The large size of the dysferlin gene poses a challenge for LGMD2B gene therapy efforts aimed at restoring dysferlin expression in skeletal muscle fibers. Here, we present an alternative gene therapy approach targeting reduced ASM secretion, the consequence of dysferlin deficit. We showed that the bulk endocytic ability is compromised in LGMD2B patient cells, which was addressed by extracellularly treating cells with ASM. Expression of secreted human ASM (hASM) using a liver-specific adeno-associated virus (AAV) vector restored membrane repair capacity of patient cells to healthy levels. A single in vivo dose of hASM-AAV in the LGMD2B mouse model restored myofiber repair capacity, enabling efficient recovery of myofibers from focal or lengthening contraction-induced injury. hASM-AAV treatment was safe, attenuated fibro-fatty muscle degeneration, increased myofiber size, and restored muscle strength, similar to dysferlin gene therapy. These findings elucidate the role of ASM in dysferlin-mediated plasma membrane repair and to our knowledge offer the first non-muscle-targeted gene therapy for LGMD2B.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Sen Chandra Sreetama
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Goutam Chandra
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Robin Ziegler
- Rare and Neurologic Diseases Research, Sanofi, Framingham, Massachusetts, USA
| | - Kanneboyina Nagaraju
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, New York, USA
| | | | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
81
|
Ballouhey O, Courrier S, Kergourlay V, Gorokhova S, Cerino M, Krahn M, Lévy N, Bartoli M. The Dysferlin Transcript Containing the Alternative Exon 40a is Essential for Myocyte Functions. Front Cell Dev Biol 2021; 9:754555. [PMID: 34888307 PMCID: PMC8650162 DOI: 10.3389/fcell.2021.754555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Dysferlinopathies are a group of muscular dystrophies caused by recessive mutations in the DYSF gene encoding the dysferlin protein. Dysferlin is a transmembrane protein involved in several muscle functions like T-tubule maintenance and membrane repair. In 2009, a study showed the existence of fourteen dysferlin transcripts generated from alternative splicing. We were interested in dysferlin transcripts containing the exon 40a, and among them the transcript 11 which contains all the canonical exons and exon 40a. This alternative exon encodes a protein region that is cleaved by calpains during the muscle membrane repair mechanism. Firstly, we tested the impact of mutations in exon 40a on its cleavability by calpains. We showed that the peptide encoded by the exon 40a domain is resistant to mutations and that calpains cleaved dysferlin in the first part of DYSF exon 40a. To further explore the implication of this transcript in cell functions, we performed membrane repair, osmotic shock, and transferrin assay. Our results indicated that dysferlin transcript 11 is a key factor in the membrane repair process. Moreover, dysferlin transcript 11 participates in other cell functions such as membrane protection and vesicle trafficking. These results support the need to restore the dysferlin transcript containing the alternative exon 40a in patients affected with dysferlinopathy.
Collapse
Affiliation(s)
| | | | | | - Svetlana Gorokhova
- INSERM, MMG, U1251, Aix Marseille University, Marseille, France.,AP-HM, Département de Génétique Médicale, Hôpital d'Enfants de la Timone, Marseille, France
| | - Mathieu Cerino
- INSERM, MMG, U1251, Aix Marseille University, Marseille, France.,AP-HM, Département de Génétique Médicale, Hôpital d'Enfants de la Timone, Marseille, France
| | - Martin Krahn
- INSERM, MMG, U1251, Aix Marseille University, Marseille, France.,AP-HM, Département de Génétique Médicale, Hôpital d'Enfants de la Timone, Marseille, France
| | - Nicolas Lévy
- INSERM, MMG, U1251, Aix Marseille University, Marseille, France.,AP-HM, Département de Génétique Médicale, Hôpital d'Enfants de la Timone, Marseille, France.,GIPTIS, Genetics Institute for Patients Therapies Innovation and Science, Marseille, France
| | - Marc Bartoli
- INSERM, MMG, U1251, Aix Marseille University, Marseille, France
| |
Collapse
|
82
|
Moshe-Lilie O, Ghetie D, Banks G, Hansford BG, Chahin N. Unusual cases of Anti-SRP necrotizing myopathy with predominant distal leg weakness and atrophy. Neuromuscul Disord 2021; 32:170-175. [PMID: 35031192 DOI: 10.1016/j.nmd.2021.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/18/2021] [Accepted: 11/14/2021] [Indexed: 11/16/2022]
Abstract
Anti-SRP necrotizing myopathy is classically characterized by subacute or chronic, severe, progressive and symmetric myositis which predominantly affects proximal muscles. We report two unusual cases presenting with predominantly distal, asymmetric weakness, with selective involvement of the posterior compartment of the thighs, gastrocnemius, and soleus muscles, in addition to inflammation and edema on STIR or T2-weighted, fat-saturated MRI. In each case, creatine kinase (CK) levels were >10 times normal and myositis panels returned positive for anti-SRP. ANA, ENA, RF, and HMGCR antibody were all negative. Nerve conduction study (NCS) was normal. Electromyography (EMG) confirmed diffuse myopathy with fibrillation potentials and positive sharp waves. Additional work up, including whole exome sequencing (WES), immunohistochemical staining for different types of muscular dystrophy, and western blot for calpain 3 and dysferlin were negative. The strength and CK levels of both patients markedly improved following immunosuppression. Our cases emphasize the importance of considering anti-SRP necrotizing myopathy in patients presenting with recent onset predominant asymmetric distal leg weakness of unclear etiology, and support the usefulness of MRI of the distal legs for early recognition. Given the potential consequences of delays in treatment of this condition, the recognition of this clinical pattern is important and can allow for prompt initiation of aggressive immunotherapies.
Collapse
Affiliation(s)
- Orly Moshe-Lilie
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Daniela Ghetie
- Department of Rheumatology, Oregon Health & Science University, Portland, OR, United States
| | - George Banks
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Barry G Hansford
- Department of Radiology, Oregon Health & Science University, Portland, OR, United States
| | - Nizar Chahin
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
83
|
Molecular and cellular basis of genetically inherited skeletal muscle disorders. Nat Rev Mol Cell Biol 2021; 22:713-732. [PMID: 34257452 PMCID: PMC9686310 DOI: 10.1038/s41580-021-00389-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
Neuromuscular disorders comprise a diverse group of human inborn diseases that arise from defects in the structure and/or function of the muscle tissue - encompassing the muscle cells (myofibres) themselves and their extracellular matrix - or muscle fibre innervation. Since the identification in 1987 of the first genetic lesion associated with a neuromuscular disorder - mutations in dystrophin as an underlying cause of Duchenne muscular dystrophy - the field has made tremendous progress in understanding the genetic basis of these diseases, with pathogenic variants in more than 500 genes now identified as underlying causes of neuromuscular disorders. The subset of neuromuscular disorders that affect skeletal muscle are referred to as myopathies or muscular dystrophies, and are due to variants in genes encoding muscle proteins. Many of these proteins provide structural stability to the myofibres or function in regulating sarcolemmal integrity, whereas others are involved in protein turnover, intracellular trafficking, calcium handling and electrical excitability - processes that ensure myofibre resistance to stress and their primary activity in muscle contraction. In this Review, we discuss how defects in muscle proteins give rise to muscle dysfunction, and ultimately to disease, with a focus on pathologies that are most common, best understood and that provide the most insight into muscle biology.
Collapse
|
84
|
Thiruvengadam G, Sreetama SC, Charton K, Hogarth M, Novak JS, Suel-Petat L, Chandra G, Allard B, Richard I, Jaiswal JK. Anoctamin 5 Knockout Mouse Model Recapitulates LGMD2L Muscle Pathology and Offers Insight Into in vivo Functional Deficits. J Neuromuscul Dis 2021; 8:S243-S255. [PMID: 34633328 PMCID: PMC8673513 DOI: 10.3233/jnd-210720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the Anoctamin 5 (Ano5) gene that result in the lack of expression or function of ANO5 protein, cause Limb Girdle Muscular Dystrophy (LGMD) 2L/R12, and Miyoshi Muscular Dystrophy (MMD3). However, the dystrophic phenotype observed in patient muscles is not uniformly recapitulated by ANO5 knockout in animal models of LGMD2L. Here we describe the generation of a mouse model of LGMD2L generated by targeted out-of-frame deletion of the Ano5 gene. This model shows progressive muscle loss, increased muscle weakness, and persistent bouts of myofiber regeneration without chronic muscle inflammation, which recapitulates the mild to moderate skeletal muscle dystrophy reported in the LGMD2L patients. We show that these features of ANO5 deficient muscle are not associated with a change in the calcium-activated sarcolemmal chloride channel activity or compromised in vivo regenerative myogenesis. Use of this mouse model allows conducting in vivo investigations into the functional role of ANO5 in muscle health and for preclinical therapeutic development for LGMD2L.
Collapse
Affiliation(s)
- Girija Thiruvengadam
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Sen Chandra Sreetama
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Karine Charton
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Marshall Hogarth
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - James S Novak
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington DC
| | - Laurence Suel-Petat
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Goutam Chandra
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGene, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Lyon, France
| | - Isabelle Richard
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jyoti K Jaiswal
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington DC
| |
Collapse
|
85
|
Ayagama T, Bose SJ, Capel RA, Priestman DA, Berridge G, Fischer R, Galione A, Platt FM, Kramer H, Burton RA. A modified density gradient proteomic-based method to analyze endolysosomal proteins in cardiac tissue. iScience 2021; 24:102949. [PMID: 34466782 PMCID: PMC8384914 DOI: 10.1016/j.isci.2021.102949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/04/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
The importance of lysosomes in cardiac physiology and pathology is well established, and evidence for roles in calcium signaling is emerging. We describe a label-free proteomics method suitable for small cardiac tissue biopsies based on density-separated fractionation, which allows study of endolysosomal (EL) proteins. Density gradient fractions corresponding to tissue lysate; sarcoplasmic reticulum (SR), mitochondria (Mito) (1.3 g/mL); and EL with negligible contamination from SR or Mito (1.04 g/mL) were analyzed using Western blot, enzyme activity assay, and liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis (adapted discontinuous Percoll and sucrose differential density gradient). Kyoto Encyclopedia of Genes and Genomes, Reactome, Panther, and Gene Ontology pathway analysis showed good coverage of RAB proteins and lysosomal cathepsins (including cardiac-specific cathepsin D) in the purified EL fraction. Significant EL proteins recovered included catalytic activity proteins. We thus present a comprehensive protocol and data set of guinea pig atrial EL organelle proteomics using techniques also applicable for non-cardiac tissue.
Collapse
Affiliation(s)
- Thamali Ayagama
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Samuel J. Bose
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Rebecca A. Capel
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | | | - Georgina Berridge
- Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Roman Fischer
- Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Antony Galione
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Frances M. Platt
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Holger Kramer
- Biological Mass Spectrometry and Proteomics Facility, MRC London Institute of Medical Sciences, Imperial College London, London, W12 0NN UK
| | | |
Collapse
|
86
|
Playfoot CJ, Duc J, Sheppard S, Dind S, Coudray A, Planet E, Trono D. Transposable elements and their KZFP controllers are drivers of transcriptional innovation in the developing human brain. Genome Res 2021; 31:1531-1545. [PMID: 34400477 PMCID: PMC8415367 DOI: 10.1101/gr.275133.120] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 07/15/2021] [Indexed: 11/25/2022]
Abstract
Transposable elements (TEs) account for more than 50% of the human genome and many have been co-opted throughout evolution to provide regulatory functions for gene expression networks. Several lines of evidence suggest that these networks are fine-tuned by the largest family of TE controllers, the KRAB-containing zinc finger proteins (KZFPs). One tissue permissive for TE transcriptional activation (termed "transposcription") is the adult human brain, however comprehensive studies on the extent of this process and its potential contribution to human brain development are lacking. To elucidate the spatiotemporal transposcriptome of the developing human brain, we have analyzed two independent RNA-seq data sets encompassing 16 brain regions from eight weeks postconception into adulthood. We reveal a distinct KZFP:TE transcriptional profile defining the late prenatal to early postnatal transition, and the spatiotemporal and cell type-specific activation of TE-derived alternative promoters driving the expression of neurogenesis-associated genes. Long-read sequencing confirmed these TE-driven isoforms as significant contributors to neurogenic transcripts. We also show experimentally that a co-opted antisense L2 element drives temporal protein relocalization away from the endoplasmic reticulum, suggestive of novel TE dependent protein function in primate evolution. This work highlights the widespread dynamic nature of the spatiotemporal KZFP:TE transcriptome and its importance throughout TE mediated genome innovation and neurotypical human brain development. To facilitate interactive exploration of these spatiotemporal gene and TE expression dynamics, we provide the "Brain TExplorer" web application freely accessible for the community.
Collapse
Affiliation(s)
- Christopher J Playfoot
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Julien Duc
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Shaoline Sheppard
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sagane Dind
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Alexandre Coudray
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Evarist Planet
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Didier Trono
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
87
|
Ataluren-Promising Therapeutic Premature Termination Codon Readthrough Frontrunner. Pharmaceuticals (Basel) 2021; 14:ph14080785. [PMID: 34451881 PMCID: PMC8398184 DOI: 10.3390/ph14080785] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 02/08/2023] Open
Abstract
Around 12% of hereditary disease-causing mutations are in-frame nonsense mutations. The expression of genes containing nonsense mutations potentially leads to the production of truncated proteins with residual or virtually no function. However, the translation of transcripts containing premature stop codons resulting in full-length protein expression can be achieved using readthrough agents. Among them, only ataluren was approved in several countries to treat nonsense mutation Duchenne muscular dystrophy (DMD) patients. This review summarizes ataluren’s journey from its identification, via first in vitro activity experiments, to clinical trials in DMD, cystic fibrosis, and aniridia. Additionally, data on its pharmacokinetics and mechanism of action are presented. The range of diseases with underlying nonsense mutations is described for which ataluren therapy seems to be promising. What is more, experiments in which ataluren did not show its readthrough activity are also included, and reasons for their failures are discussed.
Collapse
|
88
|
Barefield DY, Sell JJ, Tahtah I, Kearns SD, McNally EM, Demonbreun AR. Loss of dysferlin or myoferlin results in differential defects in excitation-contraction coupling in mouse skeletal muscle. Sci Rep 2021; 11:15865. [PMID: 34354129 PMCID: PMC8342512 DOI: 10.1038/s41598-021-95378-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022] Open
Abstract
Muscular dystrophies are disorders characterized by progressive muscle loss and weakness that are both genotypically and phenotypically heterogenous. Progression of muscle disease arises from impaired regeneration, plasma membrane instability, defective membrane repair, and calcium mishandling. The ferlin protein family, including dysferlin and myoferlin, are calcium-binding, membrane-associated proteins that regulate membrane fusion, trafficking, and tubule formation. Mice lacking dysferlin (Dysf), myoferlin (Myof), and both dysferlin and myoferlin (Fer) on an isogenic inbred 129 background were previously demonstrated that loss of both dysferlin and myoferlin resulted in more severe muscle disease than loss of either gene alone. Furthermore, Fer mice had disordered triad organization with visibly malformed transverse tubules and sarcoplasmic reticulum, suggesting distinct roles of dysferlin and myoferlin. To assess the physiological role of disorganized triads, we now assessed excitation contraction (EC) coupling in these models. We identified differential abnormalities in EC coupling and ryanodine receptor disruption in flexor digitorum brevis myofibers isolated from ferlin mutant mice. We found that loss of dysferlin alone preserved sensitivity for EC coupling and was associated with larger ryanodine receptor clusters compared to wildtype myofibers. Loss of myoferlin alone or together with a loss of dysferlin reduced sensitivity for EC coupling, and produced disorganized and smaller ryanodine receptor cluster size compared to wildtype myofibers. These data reveal impaired EC coupling in Myof and Fer myofibers and slightly potentiated EC coupling in Dysf myofibers. Despite high homology, dysferlin and myoferlin have differential roles in regulating sarcotubular formation and maintenance resulting in unique impairments in calcium handling properties.
Collapse
Affiliation(s)
- David Y Barefield
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA.
| | - Jordan J Sell
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Ibrahim Tahtah
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Samuel D Kearns
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. .,Center for Genetic Medicine, Northwestern University, 303 E Superior Lurie 5-512, Chicago, IL, 60611, USA.
| |
Collapse
|
89
|
Ganchinho Lucas S, Vieira Santos I, Pencas Alfaiate FJ, Lino I. A new dysferlin gene mutation in a Portuguese family with Miyoshi myopathy. BMJ Case Rep 2021; 14:14/7/e242341. [PMID: 34281941 DOI: 10.1136/bcr-2021-242341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Dysferlinopathies are autosomal recessive muscular dystrophies caused by mutations in the dysferlin gene (DYSF). A 33-year-old man was born to a non-consanguineous couple. At the age of 25 he stared to feel weakness of the distal lower limbs and also experienced episodes of rhabdomyolysis. Electromyography showed a myopathic pattern, and muscle biopsy revealed dystrophic changes with absence of dysferlin. Genetic analysis was positive for a mutation in the c3367_3368del DYSF gene (p.Lys1123GLUFS*2). After 8 years of disease evolution the symptomatology worsened. This is the first report of this mutation of the DYSF gene identified in a non-consanguineous Portuguese family, studied over 8 years. We believe the mutation is responsible for the Miyoshi myopathy. Disease progression cannot be predicted in either the patient or carrier family because there are no similar cases previously described in the literature.
Collapse
Affiliation(s)
| | | | | | - Ireneia Lino
- Unidade de Hospitalização Domiciliária Polivalente, Hospital do Espírito Santo EPE, Évora, Portugal
| |
Collapse
|
90
|
Annexins and Membrane Repair Dysfunctions in Muscular Dystrophies. Int J Mol Sci 2021; 22:ijms22105276. [PMID: 34067866 PMCID: PMC8155887 DOI: 10.3390/ijms22105276] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
Muscular dystrophies constitute a group of genetic disorders that cause weakness and progressive loss of skeletal muscle mass. Among them, Miyoshi muscular dystrophy 1 (MMD1), limb girdle muscular dystrophy type R2 (LGMDR2/2B), and LGMDR12 (2L) are characterized by mutation in gene encoding key membrane-repair protein, which leads to severe dysfunctions in sarcolemma repair. Cell membrane disruption is a physiological event induced by mechanical stress, such as muscle contraction and stretching. Like many eukaryotic cells, muscle fibers possess a protein machinery ensuring fast resealing of damaged plasma membrane. Members of the annexins A (ANXA) family belong to this protein machinery. ANXA are small soluble proteins, twelve in number in humans, which share the property of binding to membranes exposing negatively-charged phospholipids in the presence of calcium (Ca2+). Many ANXA have been reported to participate in membrane repair of varied cell types and species, including human skeletal muscle cells in which they may play a collective role in protection and repair of the sarcolemma. Here, we discuss the participation of ANXA in membrane repair of healthy skeletal muscle cells and how dysregulation of ANXA expression may impact the clinical severity of muscular dystrophies.
Collapse
|
91
|
Gu H, Peng Y, Chen Y. An Emerging Therapeutic Approach by Targeting Myoferlin (MYOF) for Malignant Tumors. Curr Top Med Chem 2021; 20:1509-1515. [PMID: 32552653 DOI: 10.2174/1568026620666200618123436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/13/2020] [Accepted: 06/13/2020] [Indexed: 12/31/2022]
Abstract
Myoferlin (MYOF), as a member of the ferlin family, is a type II transmembrane protein with a single transmembrane domain at the carbon terminus. Studies have shown that MYOF is involved in pivotal physiological functions related to numerous cell membranes, such as extracellular secretion, endocytosis cycle, vesicle trafficking, membrane repair, membrane receptor recycling, and secreted protein efflux. Recently, the studies have also revealed that MYOF is overexpressed in a variety of cancers such as colorectal cancer, pancreatic cancer, breast cancer, melanoma, gastric cancer, and non-small-cell lung cancer. High expression of MYOF is associated with the high invasion of tumors and poor clinical prognosis. MYOF medicates the expression, secretion, and distribution of proteins, which were closely related to cancers, as well as the energy utilization of cancer cells, lipid metabolism and other physiological activities by regulating the physiological processes of membrane transport. In this short article, we briefly summarize the latest progress related to MYOF, indicating that small molecule inhibitors targeting the MYOF-C2D domain can selectively inhibit the proliferation and migration of cancer cells, and MYOF may be a promising target for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Haijun Gu
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yangrui Peng
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
92
|
Retrospective analysis and reclassification of DYSF variants in a large French series of dysferlinopathy patients. Genet Med 2021; 23:1574-1577. [PMID: 33927379 DOI: 10.1038/s41436-021-01164-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Recent evolution of sequencing technologies and the development of international standards in variant interpretation have profoundly changed the diagnostic approaches in clinical genetics. As a consequence, many variants that were initially claimed to be disease-causing can be now reclassified as benign or uncertain in light of the new data available. Unfortunately, the misclassified variants are still present in the scientific literature and variant databases, greatly interfering with interpretation of diagnostic sequencing results. Despite the urgent need, large-scale efforts to update the classifications of these variants are still not sufficient. METHODS We retrospectively analyzed 176 DYSF gene variants that were identified in dysferlinopathy patients referred to the Marseille Medical Genetics Department for diagnostic sequencing since 2001. RESULTS We reclassified all variants into five-tier American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) pathogenicity classes, revealing changed pathogenicity for 17 variants. We then updated the information for the variants that have been previously published in the variant database and submitted 46 additional DYSF variants. CONCLUSION Besides direct benefit for dysferlinopathy diagnostics, our study contributes to the much needed effort to reanalyze variants from previously published cohorts and to work with curators of variant databases to update the entries for erroneously classified variants.
Collapse
|
93
|
Kagita A, Lung MSY, Xu H, Kita Y, Sasakawa N, Iguchi T, Ono M, Wang XH, Gee P, Hotta A. Efficient ssODN-Mediated Targeting by Avoiding Cellular Inhibitory RNAs through Precomplexed CRISPR-Cas9/sgRNA Ribonucleoprotein. Stem Cell Reports 2021; 16:985-996. [PMID: 33711268 PMCID: PMC8072016 DOI: 10.1016/j.stemcr.2021.02.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 10/25/2022] Open
Abstract
Combined with CRISPR-Cas9 technology and single-stranded oligodeoxynucleotides (ssODNs), specific single-nucleotide alterations can be introduced into a targeted genomic locus in induced pluripotent stem cells (iPSCs); however, ssODN knockin frequency is low compared with deletion induction. Although several Cas9 transduction methods have been reported, the biochemical behavior of CRISPR-Cas9 nuclease in mammalian cells is yet to be explored. Here, we investigated intrinsic cellular factors that affect Cas9 cleavage activity in vitro. We found that intracellular RNA, but not DNA or protein fractions, inhibits Cas9 from binding to single guide RNA (sgRNA) and reduces the enzymatic activity. To prevent this, precomplexing Cas9 and sgRNA before delivery into cells can lead to higher genome editing activity compared with Cas9 overexpression approaches. By optimizing electroporation parameters of precomplexed ribonucleoprotein and ssODN, we achieved efficiencies of single-nucleotide correction as high as 70% and loxP insertion up to 40%. Finally, we could replace the HLA-C1 allele with the C2 allele to generate histocompatibility leukocyte antigen custom-edited iPSCs.
Collapse
Affiliation(s)
- Akihiro Kagita
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mandy S Y Lung
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Huaigeng Xu
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuto Kita
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Noriko Sasakawa
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takahiro Iguchi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Miyuki Ono
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Xiou H Wang
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Peter Gee
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
94
|
Fernández-Eulate G, Querin G, Moore U, Behin A, Masingue M, Bassez G, Leonard-Louis S, Laforêt P, Maisonobe T, Merle PE, Spinazzi M, Solé G, Kuntzer T, Bedat-Millet AL, Salort-Campana E, Attarian S, Péréon Y, Feasson L, Graveleau J, Nadaj-Pakleza A, Leturcq F, Gorokhova S, Krahn M, Eymard B, Straub V, Evangelista T, Stojkovic T. Deep phenotyping of an international series of patients with late-onset dysferlinopathy. Eur J Neurol 2021; 28:2092-2102. [PMID: 33715265 DOI: 10.1111/ene.14821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/17/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND To describe the clinical, pathological, and molecular characteristics of late-onset (LO) dysferlinopathy patients. METHODS Retrospective series of patients with LO dysferlinopathy, defined by an age at onset of symptoms ≥30 years, from neuromuscular centers in France and the International Clinical Outcome Study for dysferlinopathy (COS). Patients with early-onset (EO) dysferlinopathy (<30 years) were randomly selected from the COS study as a control group, and the North Star Assessment for Dysferlinopathy (NSAD) and Activity Limitation (ACTIVLIM) scores were used to assess functionality. Muscle biopsies obtained from 11 LO and 11 EO patients were revisited. RESULTS Forty-eight patients with LO dysferlinopathy were included (28 females). Median age at onset of symptoms was 37 (range 30-57) years and most patients showed a limb-girdle (n = 26) or distal (n = 10) phenotype. However, compared with EO dysferlinopathy patients (n = 48), LO patients more frequently showed atypical phenotypes (7 vs. 1; p = 0.014), including camptocormia, lower creatine kinase levels (2855 vs. 4394 U/L; p = 0.01), and higher NSAD (p = 0.008) and ACTIVLIM scores (p = 0.016). Loss of ambulation in LO patients tended to occur later (23 ± 4.4 years after disease onset vs. 16.3 ± 6.8 years; p = 0.064). Muscle biopsy of LO patients more frequently showed an atypical pattern (unspecific myopathic changes) as well as significantly less necrosis regeneration and inflammation. Although LO patients more frequently showed missense variants (39.8% vs. 23.9%; p = 0.021), no differences in dysferlin protein expression were found on Western blot. CONCLUSIONS Late-onset dysferlinopathy patients show a higher frequency of atypical presentations, are less severely affected, and show milder dystrophic changes in muscle biopsy.
Collapse
Affiliation(s)
- Gorka Fernández-Eulate
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Giorgia Querin
- Plateforme I-Motion Adultes, Service de Neuromyologie, Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Ursula Moore
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Anthony Behin
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Marion Masingue
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Guillaume Bassez
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Sarah Leonard-Louis
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Pascal Laforêt
- Nord-Est/Ile-de-France Neuromuscular Reference Center, FHU PHENIX, Neurology Department, Raymond-Poincaré Hospital, Versailles Saint-Quentin-en-Yvelines - Paris Saclay University, Garches, France
| | - Thierry Maisonobe
- Department of Clinical Neurophysiology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | | | - Marco Spinazzi
- Neuromuscular Reference Center, Angers University Hospital, Angers, France
| | - Guilhem Solé
- Referral Center for Neuromuscular Diseases 'AOC', Nerve-Muscle Unit, Bordeaux University Hospitals (Pellegrin Hospital), Bordeaux, France
| | - Thierry Kuntzer
- Nerve-Muscle Unit, Department of Neurosciences, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | - Emmanuelle Salort-Campana
- PACA Réunion Rhone Alpes Neuromuscular Reference Center, APHM, La Timone University Hospital, Marseille, France
| | - Shahram Attarian
- PACA Réunion Rhone Alpes Neuromuscular Reference Center, APHM, La Timone University Hospital, Marseille, France
| | - Yann Péréon
- Reference Center for Neuromuscular Diseases Atlantique-Occitanie-Caraïbes, Nantes University Hospital, Nantes, France
| | - Leonard Feasson
- Neuromuscular Reference Center, Unit of Myology, Inter-University Laboratory of Human Movement Biology, Saint-Etienne University Hospital, Saint-Étienne, France
| | - Julie Graveleau
- Neuromuscular Reference Center, Saint-Nazaire Hospital, Saint-Nazaire, France
| | - Aleksandra Nadaj-Pakleza
- Nord-Est/Ile-de-France Neuromuscular Reference Center, Department of Neurology, Strasbourg University Hospital, Strasbourg, France
| | - France Leturcq
- Genetics and Molecular Biology Laboratory, Cochin University Hospital, Paris, France
| | - Svetlana Gorokhova
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix-Marseille University, Marseille, France.,Département de Génétique Médicale, Hôpital Timone Enfants, APHM, Marseille, France
| | - Martin Krahn
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix-Marseille University, Marseille, France.,Département de Génétique Médicale, Hôpital Timone Enfants, APHM, Marseille, France
| | - Bruno Eymard
- Nord-Est/Ile-de-France Neuromuscular Reference Center, Neurology Department, Raymond-Poincaré Hospital, Sorbonne University, Garches, France
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Teresinha Evangelista
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Tanya Stojkovic
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| |
Collapse
|
95
|
Xia Q, Huang X, Huang J, Zheng Y, March ME, Li J, Wei Y. The Role of Autophagy in Skeletal Muscle Diseases. Front Physiol 2021; 12:638983. [PMID: 33841177 PMCID: PMC8027491 DOI: 10.3389/fphys.2021.638983] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is the most abundant type of tissue in human body, being involved in diverse activities and maintaining a finely tuned metabolic balance. Autophagy, characterized by the autophagosome–lysosome system with the involvement of evolutionarily conserved autophagy-related genes, is an important catabolic process and plays an essential role in energy generation and consumption, as well as substance turnover processes in skeletal muscles. Autophagy in skeletal muscles is finely tuned under the tight regulation of diverse signaling pathways, and the autophagy pathway has cross-talk with other pathways to form feedback loops under physiological conditions and metabolic stress. Altered autophagy activity characterized by either increased formation of autophagosomes or inhibition of lysosome-autophagosome fusion can lead to pathological cascades, and mutations in autophagy genes and deregulation of autophagy pathways have been identified as one of the major causes for a variety of skeleton muscle disorders. The advancement of multi-omics techniques enables further understanding of the molecular and biochemical mechanisms underlying the role of autophagy in skeletal muscle disorders, which may yield novel therapeutic targets for these disorders.
Collapse
Affiliation(s)
- Qianghua Xia
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Xubo Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jieru Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yongfeng Zheng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Michael E March
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jin Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yongjie Wei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
96
|
Yang R, Hao Q, Lu Q, Meng F, Niu J, Liu Z, Niu G, Yu X. Fabrication of small-structure red-emissive fluorescent probes for plasma membrane enables quantification of nuclear to cytoplasmic ratio in live cells and tissues. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 249:119338. [PMID: 33360060 DOI: 10.1016/j.saa.2020.119338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 06/12/2023]
Abstract
Nuclear to cytoplasmic ratio is one of the vital parameters in diagnosis of cancer by means of hematoxylin-eosin (HE) stained histopathology. However, HE histopathology dependent on mechanical tissue slice damages biosamples and exhibits insufficient accuracy. Herein, we rationally prepared two small-molecule plasma membrane fluorescent probes with red-emitting fluorescence for visualizing plasma membrane in living cells and tissues. Their fluorescence intensities are strongly affected by environmental viscosity, which enables the exclusive imaging of plasma membrane in high fidelity. The probes can visualize plasma membrane in SiHa and rat blood red cells. Particularly, the probes are able to visualize T-tubule (transverse tubule) in skeletal muscle tissues successfully, suggesting their ability to image plasma membrane in tissues. In cooperation with Hoechst 33342, the nuclear to cytoplasmic ratio was successfully qualified in live cells and tissues. We believe these probes may have potential applications in facilitating the study on histopathology and the related areas.
Collapse
Affiliation(s)
- Rui Yang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China
| | - Qiuhua Hao
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China
| | - Qing Lu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China
| | - Fangfang Meng
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China
| | - Jie Niu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China
| | - Zhiqiang Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China.
| | - Guangle Niu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China.
| | - Xiaoqiang Yu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, PR China; Advanced Medical Research Institute, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
97
|
Foltz SJ, Cui YY, Choo HJ, Hartzell HC. ANO5 ensures trafficking of annexins in wounded myofibers. J Cell Biol 2021; 220:e202007059. [PMID: 33496727 PMCID: PMC7844426 DOI: 10.1083/jcb.202007059] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/20/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in ANO5 (TMEM16E) cause limb-girdle muscular dystrophy R12. Defective plasma membrane repair is a likely mechanism. Using myofibers from Ano5 knockout mice, we show that trafficking of several annexin proteins, which together form a cap at the site of injury, is altered upon loss of ANO5. Annexin A2 accumulates at the wound to nearly twice the level observed in WT fibers, while annexin A6 accumulation is substantially inhibited in the absence of ANO5. Appearance of annexins A1 and A5 at the cap is likewise diminished in the Ano5 knockout. These changes are correlated with an alteration in annexin repair cap fine structure and shedding of annexin-positive vesicles. We conclude that loss of annexin coordination during repair is disrupted in Ano5 knockout mice and underlies the defective repair phenotype. Although ANO5 is a phospholipid scramblase, abnormal repair is rescued by overexpression of a scramblase-defective ANO5 mutant, suggesting a novel, scramblase-independent role of ANO5 in repair.
Collapse
Affiliation(s)
| | | | - Hyojung J. Choo
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - H. Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
98
|
Gupta S, Yano J, Mercier V, Htwe HH, Shin HR, Rademaker G, Cakir Z, Ituarte T, Wen KW, Kim GE, Zoncu R, Roux A, Dawson DW, Perera RM. Lysosomal retargeting of Myoferlin mitigates membrane stress to enable pancreatic cancer growth. Nat Cell Biol 2021; 23:232-242. [PMID: 33686253 PMCID: PMC9446896 DOI: 10.1038/s41556-021-00644-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Lysosomes must maintain the integrity of their limiting membrane to ensure efficient fusion with incoming organelles and degradation of substrates within their lumen. Pancreatic cancer cells upregulate lysosomal biogenesis to enhance nutrient recycling and stress resistance, but it is unknown whether dedicated programmes for maintaining the integrity of the lysosome membrane facilitate pancreatic cancer growth. Using proteomic-based organelle profiling, we identify the Ferlin family plasma membrane repair factor Myoferlin as selectively and highly enriched on the membrane of pancreatic cancer lysosomes. Mechanistically, lysosomal localization of Myoferlin is necessary and sufficient for the maintenance of lysosome health and provides an early acting protective system against membrane damage that is independent of the endosomal sorting complex required for transport (ESCRT)-mediated repair network. Myoferlin is upregulated in human pancreatic cancer, predicts poor survival and its ablation severely impairs lysosome function and tumour growth in vivo. Thus, retargeting of plasma membrane repair factors enhances the pro-oncogenic activities of the lysosome.
Collapse
Affiliation(s)
- Suprit Gupta
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Julian Yano
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Vincent Mercier
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Htet Htwe Htwe
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Hijai R Shin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Gilles Rademaker
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Zeynep Cakir
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Ituarte
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Kwun W Wen
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Grace E Kim
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - David W Dawson
- Department of Pathology and Laboratory Medicine and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Rushika M Perera
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
99
|
Moore U, Gordish H, Diaz-Manera J, James MK, Mayhew AG, Guglieri M, Fernandez-Torron R, Rufibach LE, Feng J, Blamire AM, Carlier PG, Spuler S, Day JW, Jones KJ, Bharucha-Goebel DX, Salort-Campana E, Pestronk A, Walter MC, Paradas C, Stojkovic T, Mori-Yoshimura M, Bravver E, Pegoraro E, Lowes LP, Mendell JR, Bushby K, Straub V. Miyoshi myopathy and limb girdle muscular dystrophy R2 are the same disease. Neuromuscul Disord 2021; 31:265-280. [PMID: 33610434 DOI: 10.1016/j.nmd.2021.01.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 10/22/2022]
Abstract
This study aims to determine clinically relevant phenotypic differences between the two most common phenotypic classifications in dysferlinopathy, limb girdle muscular dystrophy R2 (LGMDR2) and Miyoshi myopathy (MMD1). LGMDR2 and MMD1 are reported to involve different muscles, with LGMDR2 showing predominant limb girdle weakness and MMD1 showing predominant distal lower limb weakness. We used heatmaps, regression analysis and principle component analysis of functional and Magnetic Resonance Imaging data to perform a cross-sectional review of the pattern of muscle involvement in 168 patients from the Jain Foundation's international Clinical Outcomes Study for Dysferlinopathy. We demonstrated that there is no clinically relevant difference in proximal vs distal involvement between diagnosis. There is a continuum of distal involvement at any given degree of proximal involvement and patients do not fall into discrete distally or proximally affected groups. There appeared to be geographical preference for a particular diagnosis, with MMD1 being more common in Japan and LGMDR2 in Europe and the USA. We conclude that the dysferlinopathies do not form two distinct phenotypic groups and therefore should not be split into separate cohorts of LGMDR2 and MM for the purposes of clinical management, enrolment in clinical trials or access to subsequent treatments.
Collapse
Affiliation(s)
- Ursula Moore
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Heather Gordish
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, United States; Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, DC, United States
| | - Jordi Diaz-Manera
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain; Neuromuscular Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Anna G Mayhew
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Roberto Fernandez-Torron
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom
| | | | - Jia Feng
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, United States
| | - Andrew M Blamire
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, United Kingdom
| | - Pierre G Carlier
- AIM & CEA NMR Laboratory, Institute of Myology, Pitié-Salpêtrière University Hospital, 47-83 Paris, France
| | - Simone Spuler
- Charite Muscle Research Unit, Experimental and Clinical Research Center, a Joint Cooperation of the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - John W Day
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Kristi J Jones
- The Children's Hospital at Westmead, and The University of Sydney, Australia
| | - Diana X Bharucha-Goebel
- Department of Neurology Children's National Health System, Washington, DC, United States; National Institutes of Health (NINDS), Bethesda, MD, United States
| | | | - Alan Pestronk
- Department of Neurology Washington University School of Medicine, St. Louis, MO, United States
| | - Maggie C Walter
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Germany
| | - Carmen Paradas
- Neuromuscular Unit, Department of Neurology, Hospital U. Virgen del Rocío/Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Tanya Stojkovic
- Centre de référence des maladies neuromusculaires, Institut de Myologie, AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry Tokyo, Japan
| | - Elena Bravver
- Neuroscience Institute, Carolinas Neuromuscular/ALS-MDA Center, Carolinas HealthCare System, Charlotte, NC, United States
| | | | - Linda Pax Lowes
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH United States
| | - Jerry R Mendell
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH United States
| | - Kate Bushby
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Central Parkway, Newcastle upon Tyne, United Kingdom.
| | | |
Collapse
|
100
|
Zhang H, Li Y, Cheng Q, Chen X, Yu Q, Li Z. Abnormal Expression of Dysferlin in Blood Monocytes Supports Primary Dysferlinopathy in Patients Confirmed by Genetic Analyses. Front Neurol 2021; 11:540098. [PMID: 33613410 PMCID: PMC7890250 DOI: 10.3389/fneur.2020.540098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: Dysferlin deficiency causes dysferlinopathy. This study aimed to expand the mutational spectrum of dysferlinopathies, to further study one case with diagnostic ambiguity, and to identify the diagnostic value of dysferlin expression in total peripheral blood mononuclear cells (PBMC). Methods: The clinical and molecular profiles of dysferlinopathies in eight Chinese patients were evaluated. We also conducted magnetic resonance imaging (6/8) and determined dysferlin protein expression in muscle (7/8) and PBMC (3/8). Results: Nine of the 13 DYSF mutations identified were novel. One patient was homozygous for the Gln111Ter mutation by genomic DNA sequencing but was found to be heterozygous by sequencing of cDNA from total PBMC. A daughter of this patient did not carry any Gln111Ter mutation. Abnormal muscle MRI with predominant involvement of the medial gastrocnemius and soleus muscle was observed in 5/6 patients. Dysferlin levels were significantly reduced (immunohistochemistry/immunoblot) or absent (immunohistochemistry) in muscle and total PBMC (26–39%) for most patients. Sarcoplasmic accumulation of dysferlin was detected in one patient. Conclusion: Genomic DNA sequencing detects frequent homozygous mutations, while fewer heterozygous mutations in cDNA are detected after posttranscription. Total PBMC may serve as an alternative to confirm diagnosis and to guide further testing in dysferlinopathies. Our results contribute to the mutational spectrum of dysferlinopathies.
Collapse
Affiliation(s)
- Huili Zhang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yaqin Li
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qiusheng Cheng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xi Chen
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiuxia Yu
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ze Li
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|